Loading

Commentary Open Access
Volume 2 | Issue 4 | DOI: https://doi.org/10.33696/immunology.2.032

CRISPR Taking the Front Seat in Immunotherapy

  • 1T Cell Engineering, Mayo Clinic, Rochester, MN, USA
  • 2Division of Hematology, Mayo Clinic, Rochester, MN, USA
  • 3Department of Immunology, Mayo Clinic, Rochester, MN, USA
  • 4Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
  • 5Division of Infectious Diseases, Mayo Clinic, Rochester, MN, USA
+ Affiliations - Affiliations

*Corresponding Author

Andrew Badley, badley.andrew@mayo.edu

Received Date: February 07, 2020

Accepted Date: May 04, 2020

Introduction

CRISPR (clustered regularly interspaced short palindromic repeats) technology has dramatically simplified genome editing and is widely applicable in both basic research and therapeutic areas. The basic principle of CRISPR relies on the use of guide RNA which is designed to bind to the DNA sequence of interest along with a CRISPR-associated (Cas) endonuclease which introduces a double-stranded DNA break (DSB) at that site. Prior to the advent of the CRISPR revolution within the last decade, gene editing was dependent on predicting and engineering protein-DNA interactions mediated by zinc-finger nucleases (ZFNs) and transcription activatorlike effector nucleases (TALENs). In contrast, CRISPR relies on simple Watson-Crick base pairing between the guide RNA and the target DNA sequence. The simplicity and versatility of CRISPR has led to its widespread implementation by the scientific community and subsequent unparalleled progress in genetic engineering.

Mechanisms of CRISPR Induced DNA Breaks

Since the first report of CRISPR-edited human cells in 2013 [1], CRISPR systems have been used to both knock out and knock in genes. Gene knockouts are created through the nonhomologous end-joining (NHEJ) pathway, which rapidly ligates the ends of the DSB without a DNA repair template [2]. NHEJ is error-prone and frequently introduces small indels into the gene, most often leading to frameshift mutations and loss-offunction of the target gene, although NHEJ has also been used to restore function in genes with existing frameshift mutations [3]. Alternatively, gene knock-ins can be achieved through homology-directed repair (HDR), a separate pathway which uses a DNA template with long homologous sequences to repair the DNA. Instead of using the sister chromatid as a repair template, researchers have exploited HDR to generate CRISPR-mediated gene knock-ins by including an exogenous DNA template with the desired transgene flanked by sequences homologous to either end of the DSB [3,4]. Microhomology-mediated end-joining (MMEJ) is an alternate pathway which relies on short homology arms to repair the DSB [5]. Though the mechanisms of MMEJ are not as well understood, MMEJ can also be used to knock in genes using CRISPR technology [6,7]. MMEJ-mediated knock-in strategies appear experimentally similar to those used for HDR, but the use of shorter homology arms allows for easier cloning of the knockin template [6,7].

Application of CRISPR in Immunotherapy

In their review article in the May 2019 edition of Haematologica, González-Romero et al. [8] discuss the uses of CRISPR-Cas9 systems in hematological diseases and succinctly outline the uses of CRISPR in the modification of chimeric antigen receptor T (CART) cells. CARs are synthetic receptors comprised of an extracellular single chain variable fragment of an antibody, hinge region and transmembrane domain, and intracellular signaling domains derived from the T cell receptor (TCR). The CAR redirects and activates T cells against cancer cells which express the cognate antigen. The authors briefly discussed various CRISPR applications in CART therapy and concluded that the combination of CRISPR and CART will fine-tune the engineered T cells and advance the field of cancer immunotherapy. Indeed, CRISPR-modified CART cells have shown to be safer and have better antitumor efficacy compared to traditionally manufactured CART cells in preclinical settings, and several clinical trials are underway (Table 1) [9-15].

Objective CRISPR
System
Delivery Methods Targets Reference
Universal, exhaustion- resistant Cas9 Electroporation of Cas9 mRNA and guide RNA + lentiviral CD19- or PSCA- CAR TCR, HLA, PD1 Ren et al. [22]
Universal, exhaustion- resistant Cas9 Electroporation of Cas9 mRNA + lentiviral CD19- or PSCA-CAR and guide RNA cassettes TCR, HLA, PD1, CTLA-4 Ren et al. [23]
Universal, exhaustion- resistant Cas9 Electroporation of Cas9/ guide RNA RNPs + AAV EGFRvIII-CAR TCR, HLA, PD1 Choi et al. [9]
Prevention or reduction of CRS and neurotoxicity Cas9 Electroporation of Cas9/ guide RNA RNPs + lentiviral CD19-CAR GM-CSF Sterner et al. [21]
Targeted CAR knockin Cas9 Electroporation of Cas9 mRNA and guide RNA + AAV CD19-CAR with homology arms TCR (CAR knockin) Eyquem et al. [19]
Targeted dual CAR knockin Cas12a Electroporation of Cas12a mRNA + lentiviral
CD19- or CD22-CAR with homology arms and guide RNA cassettes
TCR, PD1 (CAR
knockins)
Dai et al. [25]

Table 1: Preclinical CRISPR-edited CART studies.

Scientists have been using genome editing technology to create universal CART cells for nearly a decade. Knocking out the TCR and human leukocyte antigen (HLA) molecules are key to mitigating both the risk of graft-versus-host disease (GvHD) and the possibility of graft rejection of allogeneic cell products [10,16]. In 2012, researchers used ZFNs to knock out TCRα and TCRβ constant (TRAC and TRBC) chains in CART cells and demonstrated that these TCR-knockout CART cells only responded to stimulation through the CAR and did not cause GvHD in mice [17]. The following year, the group used the same approach to knock out the HLA-A gene to circumvent graft rejection of the engineered T cells; HLA-A-knockout T cells evaded killing by HLA-restricted T cells from a different donor and also avoided natural killer cell immunosurveillance by expressing nonclassical HLA molecules [17]. Similarly, TALEN technology was used to disrupt TRAC and CD52 genes in CART cells to prevent GvHD and to render the CART product resistant to elimination by the anti-CD52 lymphodepleting agent, alemtuzumab. This CART product was taken to the clinic and resulted in the complete remission and subsequent successful allogeneic stem cell transplantation of two infants with B-cell acute lymphoblastic leukemia [18].

Given the promising preclinical and clinical results in genome-edited CART cells, many groups have used CRISPR to knock out one or more endogenous genes in CART cells. These studies generally aim to 1) enhance CART efficacy by knocking out genes which lead to T cell exhaustion [9,13,15], 2) create an off-the-shelf allogeneic CART product by knocking out TCR and HLA components [19], 3) allow CART cells to attack malignant T cells without the risk of fratricide by knocking out the CAR target on the CART cell [20], or 4) improve CART safety profiles by knocking out genes which contribute to adverse side effects associated with CART therapy [21].

Researchers have sought to create allogeneic, exhaustion-resistant CART cells by combining lentiviral transduction of the CAR transgene with electroporation of Cas9 mRNA and guide RNA targeted to PD1, TCR, and HLA gene loci. These modified CART cells demonstrated reduced alloreactivity in vitro and improved antitumor effects in both hematological and solid tumor xenograft models which express the PD1 ligand PD-L1 [22]. The same group employed a multiplex genome editing platform by including multiple guide RNA cassettes with the CAR transgene in a lentiviral vector, allowing for simultaneous knockout of up to four genes in the CART cells after a single electroporation of Cas9 mRNA [23]. Triple knockout of PD1, TCR, and HLA molecules in CART cells also prolonged survival in a mouse glioblastoma model [9]. CRISPR-mediated knockout of checkpoint molecules such as PD1 may improve CART activity especially in solid tumors, an area in which the CART field has seen limited success in part due to the immunosuppressive tumor microenvironment. Several clinical trials in China are ongoing in testing CRISPRedited CART cells with knockout of immune checkpoint molecules such as PD1 (NCT03747965, NCT03545815) and HPK1 (NCT04037566) as well as universal CART cells (NCT03166878, NCT03398967) (Table 2). CRISPRCART clinical trials also have been initiated in the US to test allogeneic CART cells (NCT04035434) (Table 2). Until recently, CART therapy could not be used to treat cancers such as T-cell acute lymphoblastic leukemia, as the CARs would be activated against T cell antigens expressed on both the malignant T cells and the CART cells themselves. Scientists have used CRISPR to produce CD7-targeting CART cells with CD7 knocked out, rendering them cytotoxic to endogenous T cells but resistant to their own CAR; a clinical trial has been initiated in the US to test this platform in patients with a variety of T cell malignancies (NCT03690011) (Table 2).

Objective CAR Target CRISPR Target Clinical Trial
Identifier
Exhaustion-resistant Mesothelin PD1 NCT03747965
Exhaustion-resistant CD19 HPK1 NCT04037566
Exhaustion-resistant, universal Mesothelin PD1, TCR NCT03545815
Allogeneic CD19 TCR, HLA NCT04035434
Allogeneic CD19 TCR, HLA NCT03166878
Allogeneic CD19/CD20 or CD19/ CD22 TCR, HLA NCT03398967
Target T cell malignancies CD7 CD7 NCT03690011

Table 2: Initiated and ongoing clinical trials of CRISPR-edited CART cells.

Scientists have also used CRISPR-edited CART cells to prevent or reduce cytokine release syndrome (CRS) and neurotoxicity, the two most common severe side effects of CART therapy [21]. Granulocyte-macrophage colonystimulating factor (GM-CSF) is critical to monocyte modulation, which in turn plays a role in the development of both CRS and neurotoxicity. Our group has shown that GM-CSF neutralization prevents the development of CRS and leads to significant reduction of neurotoxicity. Additionally, using CRISPR-Cas9 to knock out GM-CSF in CART cells had no adverse effects on normal CART cell function, and these knockout CART cells also displayed superior anti-leukemic activity and prolonged overall survival in vivo compared to wild-type GM-CSF CART cells [14,21]. Interestingly, the improved antitumor efficacy was observed in an immunocompromised mouse model with an absence of myeloid cells, indicating direct effects of GM-CSF knockout on CART cells in addition to the impact on myeloid cells. Further analysis uncovered a link between GM-CSF knockout and inhibition of the Fas death pathway [24]. CRISPR technology has enabled the production of CART cells that are both safer and more effective in preclinical studies and presents an exciting opportunity for bringing improved CART products to patients.

Most studies to date have used CRISPR to knock out undesired genes in CART cells, but CRISPR has also been used to knock in the CAR at a specific locus in the T cell genome [19]. CRISPR-Cas9 and CAR transgenes have been combined in a lentiviral vector to knock in the CAR transgene at the TRAC locus, resulting in simultaneous TCR knockout. These CART cells had stronger antileukemic activity in a mouse xenograft model [19]. An alternate system, CRISPR-Cas12a, was used in combination with CARs in adeno-associated viral vectors to generate double knock-in/knock-outs with a CD19 CAR at the PD1 locus and a CD22 CAR at the TRAC locus [25]. Interestingly, Cas12a was more effective at producing double knock-in/knock-outs than the more commonly used CRISPR-Cas9 platform, and CRISPR-Cas12a-edited CART cells expressed lower levels of exhaustion markers than CRISPR-Cas9-edited CART cells [25]. Furthermore, targeted CAR delivery creates a more uniform cellular product, in sharp contrast with the random integration of viral transduction methods most commonly used to generate CART cells [25]. CART therapy is expensive and highly variable, and specific CAR integration into a known genomic locus will reduce batch-to-batch variation. In turn, more uniform CART products will likely diminish the regulatory cost and burden involved in producing current CART products as well as in approving new CART therapies. Insertional oncogenesis, although never observed with modified T cells in the clinic to date, remains a possibility with any therapy involving a non-targeted, integrated transgene; CAR integration at a known genomic site would alleviate these concerns as well.

As stated in the review by González-Romero et al. [8], it would be “hard to underestimate” the impact of CRISPR technology paired with CART cells. Great strides have been taken to improve both the function and safety profile of CART cells, as well as in creating universal allogeneic CART cells to overcome the logistical and economic hurdles presented by current CART manufacturing techniques [9,10,13,14,19,22,23]. However, there are many areas that need additional exploration. Cas9 is the CRISPR system most commonly implemented in genome editing, but several other CRISPR variants are also viable candidates, most notably CRISPR-Cas12a. Cas12a recognizes a different protospacer adjacent motif than Cas9, broadening the potential target sequences. Instead of creating blunt ends at the DSB as seen in Cas9, Cas12a generates sticky ends which are less likely to be repaired by NHEJ and may be preferable to HDRmediated gene knock-ins [25]. Direct comparisons of Cas9 and Cas12a systems are largely lacking to date. The majority of studies have used various viral vectors to edit the T cell genome, but when translating CART to a clinical and commercial product, GMP-grade virus presents a huge expense and logistical challenge. One report described the generation of CRISPR-edited TCRengineered T cells using completely nonviral methods [26]. More research is needed in the production of nonviral CRISPR-edited CART cells to overcome current manufacturing challenges of clinical-grade CART products. Finally, CRISPR is most often used to enhance the therapeutic efficacy of CART cells, but it can also be a valuable screening tool. One such study used CRISPR gene knockout screens to investigate the mechanisms behind the effects of various small molecule drugs on CART cytotoxicity [27]. CRISPR libraries can enhance our understanding of CART interactions with existing drugs and influence choices made in the clinic regarding CART and chemotherapeutic drug combinations. Broadening the scope from CART cells, a recent study used a genomewide CRISPR screen to discover an unconventional TCR that appears to be cancer-specific and HLA-independent [11]. Whether these unconventional T cells can be used as a standalone therapeutic or as a complement to traditional cancer treatments or alongside CART therapy remains to be seen, but this study highlights the importance of CRISPR technology in the discovery of additional immunotherapies. CRISPR has been used to enhance existing therapies, but it may also uncover entirely new avenues of cellular immunotherapy. CRISPR has proven to be a valuable tool in preclinical studies of CART cells, and clinical trials of CRISPR-edited CART cells will yield more information on utilizing this versatile genome editing technique to improve cancer immunotherapies and patient outcomes.

Conclusion

In summary, the advances in CRISPR technology over the last decade have started to revolutionize cellular immunotherapy for the treatment of cancer. We have witnessed CRISPR genome engineering moving from “proof of concept” experiments to first in human clinical trials.

Disclosures

SSK is inventor on patents in the field of CART immunotherapy that are licensed to Novartis, Humanigen, and Mettaforge. SSK receives research funding from Kite, Gilead, Juno, Celgene, Tolero, Morphosys, Novartis, Humanigen, Lentigen, and Sunesis. SSK has served on advisory boards for Kite, Juno and Humanigen. ADB is a consultant for Abbvie, is a member of advisory boards for Nference and Zentalis, and is founder and president of Splissen pharmaceuticals.

References

1. Jinek M, East A, Cheng A, Lin S, Ma E, Doudna J. RNA-programmed genome editing in human cells. Elife. 2013 Jan 29;2:e00471.

2. Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science. 346, 1258096.

3. Hoeijmakers JH. Genome maintenance mechanisms for preventing cancer. Nature. 2001 May;411(6835):366- 74.

4. Qi Z, Redding S, Lee JY, Gibb B, Kwon Y, Niu H, et al. DNA sequence alignment by microhomology sampling during homologous recombination. Cell. 2015 Feb 26;160(5):856-69.

5. Ahrabi S, Sarkar S, Pfister SX, Pirovano G, Higgins GS, Porter AC, et al. A role for human homologous recombination factors in suppressing microhomologymediated end joining. Nucleic Acids Research. 2016 Jul 8;44(12):5743-57.

6. McVey M, Lee SE. MMEJ repair of double-strand breaks (director’s cut): deleted sequences and alternative endings. Trends in Genetics. 2008 Nov 1;24(11):529-38.

7. Khodaverdian VY, Hanscom T, Yu AM, Yu TL, Mak V, Brown AJ, et al. Secondary structure forming sequences drive SD-MMEJ repair of DNA double-strand breaks. Nucleic Acids Research. 2017 Dec 15;45(22):12848-61.

8. González-Romero E, Martínez-Valiente C, García- Ruiz C, Vázquez-Manrique RP, Cervera J, Sanjuan-Pla A. CRISPR to fix bad blood: a new tool in basic and clinical hematology. Haematologica. 2019 May 1;104(5):881-93.

9. Choi BD, Yu X, Castano AP, Darr H, Henderson DB, Bouffard AA, et al. CRISPR-Cas9 disruption of PD-1 enhances activity of universal EGFRvIII CAR T cells in a preclinical model of human glioblastoma. Journal for Immunotherapy of Cancer. 2019 Dec;7(1):1-8.

10. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA, Zhang F. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013 Feb 15;339(6121):819-23.

11. Crowther MD, Dolton G, Legut M, Caillaud ME, Lloyd A, Attaf M, et al. Genome-wide CRISPR–Cas9 screening reveals ubiquitous T cell cancer targeting via the monomorphic MHC class I-related protein MR1. Nature Immunology. 2020 Feb;21(2):178-85.

12. June CH. Emerging Use of CRISPR Technology— Chasing the Elusive HIV Cure. New England Journal of Medicine. 381, 1281-1283 (2019).

13. Stadtmauer EA, Fraietta JA, Davis MM, Cohen AD, Weber KL, Lancaster E, Mangan PA, Kulikovskaya I, Gupta M, Chen F, Tian L. CRISPR-engineered T cells in patients with refractory cancer. Science. 2020 Feb 28;367(6481).

14. Sterner RM, Cox MJ, Sakemura R, Kenderian SS. Using CRISPR/Cas9 to Knock Out GM-CSF in CAR-T Cells. Journal of Visualized Experiments: JoVE. 2019 Jul(149).

15. Su S, Zou Z, Chen F, Ding N, Du J, Shao J, et al. CRISPR-Cas9-mediated disruption of PD-1 on human T cells for adoptive cellular therapies of EBV positive gastric cancer. Oncoimmunology. 2017 Jan 2;6(1):e1249558.

16. Qasim W, Amrolia PJ, Samarasinghe S, Ghorashian S, Zhan H, Stafford S, et al. First clinical application of Talen engineered universal CAR19 T cells in B-ALL. Blood 126, 2046-2046 (2015).

17. Torikai H, Reik A, Liu PQ, Zhou Y, Zhang L, Maiti S, et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimericantigen- receptor and eliminate expression of endogenous TCR. Blood, The Journal of the American Society of Hematology. 2012 Jun 14;119(24):5697-705.

18. Qasim W, Zhan H, Samarasinghe S, Adams S, Amrolia P, Stafford S, et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Science Translational Medicine. 2017 Jan 25;9(374):eaaj2013.

19. Eyquem J, Mansilla-Soto J, Giavridis T, van der Stegen SJ, Hamieh M, Cunanan KM, et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature. 2017 Mar;543(7643):113-7.

20. Gomes-Silva D, Srinivasan M, Sharma S, Lee CM, Wagner DL, Davis TH, et al. CD7-edited T cells expressing a CD7-specific CAR for the therapy of T-cell malignancies. Blood, The Journal of the American Society of Hematology. 2017 Jul 20;130(3):285-96.

21. Sterner RM, Sakemura R, Cox MJ, Yang N, Khadka RH, Forsman CL, et al. GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood, The Journal of the American Society of Hematology. 2019 Feb 14;133(7):697-709.

22. Ren J, Liu X, Fang C, Jiang S, June CH, Zhao Y. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clinical Cancer Research. 2017 May 1;23(9):2255-66.

23. Ren J, Zhang X, Liu X, Fang C, Jiang S, June CH, et al. A versatile system for rapid multiplex genome-edited CAR T cell generation. Oncotarget. 2017 Mar 7;8(10):17002.

24. Cox MJ, Kuhlmann C, Sterner RM, Sakemura R, Sinha S, Hefazi M, et al. Improved Anti-Tumor Response of Chimeric Antigen Receptor T Cell (CART) Therapy after GM-CSF Inhibition Is Mechanistically Supported By a Novel Direct Interaction of GM-CSF with Activated Carts. Biology of Blood and Marrow Transplantation. 2020 Mar 1;26(3):S60-1.

25. Dai X, Park JJ, Du Y, Kim HR, Wang G, Errami Y, et al. One-step generation of modular CAR-T cells with AAV–Cpf1. Nature Methods. 2019 Mar;16(3):247-54.

26. Roth TL, Puig-Saus C, Yu R, Shifrut E, Carnevale J, Li PJ, et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature. 2018 Jul;559(7714):405-9.

27. Brück O, Dufva O, Hohtari H, Blom S, Turkki R, Ilander M, et al. Immune profiles in acute myeloid leukemia bone marrow associate with patient age, T-cell receptor clonality, and survival. Blood Advances. 2020 Jan 28;4(2):274-86.

Author Information X